TWI435727B - Use of modulating secretion of cytokines - Google Patents

Use of modulating secretion of cytokines Download PDF

Info

Publication number
TWI435727B
TWI435727B TW98145264A TW98145264A TWI435727B TW I435727 B TWI435727 B TW I435727B TW 98145264 A TW98145264 A TW 98145264A TW 98145264 A TW98145264 A TW 98145264A TW I435727 B TWI435727 B TW I435727B
Authority
TW
Taiwan
Prior art keywords
arthritis
mangosteen
extract
tnf
mangostin
Prior art date
Application number
TW98145264A
Other languages
Chinese (zh)
Other versions
TW201121556A (en
Inventor
Lain Tze Lee
Munekatsu Iinuma
Ying Chu Shih
Shu Jiau Chiou
Chonwen Wong
Original Assignee
Ind Tech Res Inst
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ind Tech Res Inst filed Critical Ind Tech Res Inst
Priority to TW98145264A priority Critical patent/TWI435727B/en
Publication of TW201121556A publication Critical patent/TW201121556A/en
Application granted granted Critical
Publication of TWI435727B publication Critical patent/TWI435727B/en

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Description

調節細胞激素分泌之用途Use to regulate the secretion of cytokines

本發明係關於治療關節炎及調節細胞激素分泌之醫藥組成物,特別是關於以山竹萃取物為活性成分之治療關節炎及調節細胞激素分泌之醫藥組成物。The present invention relates to a pharmaceutical composition for treating arthritis and regulating cytokine secretion, and more particularly to a pharmaceutical composition for treating arthritis and regulating cytokine secretion by using mangosteen extract as an active ingredient.

由於醫療技術的進步及普及,全球人口結構趨向於高齡化,衍生出老年慢性疾病相關的公眾健康議題。關節炎向來為高齡化社會高成本支出的慢性疾病,可源自於老化、肥胖或免疫疾病等因素。傳統治療使用類固醇藥物(corticosteroids)降低發炎症狀及抑制免疫系統,或使用非類固醇藥物(NSAIDS),例如COX-1抑制劑、COX-2抑制劑、希樂葆(celebrex)、或布洛芬(ibuprofen),減少疼痛及發炎,但皆會造成嚴重程度不一的副作用。新一代治療關節炎的藥物,例如疾病調節抗風濕藥物(DMARDs),如胺基甲基葉酸(methotrexate),可減緩或停止免疫系統攻擊關節,或者生物製劑(biologics),如恩博(etanercept)、因福利美(infliximab)等,可阻斷涉及發炎過程的免疫系統中特定分子路徑,進而緩慢病程進展。但是,使用這些藥物仍必須特別注意伴隨而來的副作用。Due to the advancement and popularization of medical technology, the global population structure tends to be aging, and public health issues related to chronic diseases in the elderly are derived. Arthritis has long been a chronic disease with high cost of aging society, which can be derived from factors such as aging, obesity or immune diseases. Traditional treatments use steroidal drugs to reduce inflammatory symptoms and suppress the immune system, or use non-steroidal drugs (NSAIDS) such as COX-1 inhibitors, COX-2 inhibitors, celebrex, or ibuprofen ( Ibuprofen) reduces pain and inflammation, but it can cause side effects of varying degrees of severity. A new generation of drugs for the treatment of arthritis, such as disease-modifying anti-rheumatic drugs (DMARDs), such as aminomethyxate, can slow or stop the immune system from attacking joints, or biologics such as etanercept. Because of infliximab, etc., it can block specific molecular pathways in the immune system involved in the inflammatory process, and thus progress slowly. However, the use of these drugs must still pay special attention to the accompanying side effects.

對於關節炎的治療也逐漸轉向自然療法(natural remedy),中草藥治療是重要的一環。相關研究顯示雷公藤(Tripterygium wilfordii Hook F)(美國專利5,580,562)及柴胡(Bupleurum )(台灣專利申請案TW 97151783)具有治療關節 炎的功效。然而,開發治療效果佳且副作用少的新穎藥物仍有需要。The treatment of arthritis has also gradually turned to natural remedy, and herbal treatment is an important part. Related research shows that triptolide (Tripterygium wilfordii Hook F) (US Patent 5,580,562) and Bupleurum (Bupleurum) (Taiwan patent application TW 97151783) has the effect of treating arthritis. However, there is still a need to develop novel drugs with good therapeutic effects and few side effects.

山竹(Garcinia mangostana L.)為特產於東南亞的植物,當地的傳統醫學將山竹果皮應用於治療皮膚感染及傷口處理已行之有年。Matsumoto等人由山竹果皮中純化出α-山竹素(mangostin)、β-山竹素、γ-山竹素、及甲基-β-山竹素,並研究該化合物對細胞周期各階段的抑制作用,顯示該化合物具有抗細胞增殖效果及抗腫瘤效應(Matsumoto K.,et al.,Xanthones induce cell-cycle arrest and apoptosis in human colon cancer DLD-1 cells,Bioorg.Med.Chem .2005,13,6064-6069.)。Mangosteen ( Garcinia mangostana L.) is a plant specializing in Southeast Asia. Local traditional medicine has been used for the treatment of skin infections and wound treatment for many years. Matsumoto et al. purified α-mangostin, β-mangostin, γ-mangostin, and methyl-β-mangostin from mangosteen peel, and studied the inhibitory effect of this compound on various stages of the cell cycle. The compound has anti-cell proliferation effect and anti-tumor effect (Matsumoto K., et al., Xanthones induce cell-cycle arrest and apoptosis in human colon cancer DLD-1 cells, Bioorg. Med. Chem. 2005, 13, 6064-6069 .).

山竹亦被研究於乳癌(Moongkarndi P.,et al,Antiproliferation,antioxidation and induction of apoptosis by Garcinia mangostana(mangosteen)on SKBR3 human breast cancer cell line,J.Ethnopharmacol .2004,90(1):161-6)、抗過敏藥物(Nakatani K.,et al.,Inhibitions of histamine release and prostaglandin E2 synthesis by mangosteen,a Thai medicinal plant,Bio Pharm Bull ,2002(9):1137-41.)、及肌肉相關疾病(WO 2007/128465)等領域,亦開發作為日常生活的營養補充劑(WO 2006/060578)及化妝品(WO 2007/002666)等。Mangosteen has also been studied in breast cancer (Moongkarndi P., et al, Antiproliferation, antioxidation and induction of apoptosis by Garcinia mangostana (mangosteen) on SKBR3 human breast cancer cell line, J. Ethnopharmacol. 2004, 90(1): 161-6) , anti-allergic drugs (Nakatani K., et al., Inhibitions of histamine release and prostaglandin E2 synthesis by mangosteen, a Thai medicinal plant, Bio Pharm Bull , 2002 (9): 1137-41.), and muscle-related diseases (WO In the field of 2007/128465), it is also developed as a nutritional supplement for daily life (WO 2006/060578) and cosmetics (WO 2007/002666).

在一發明態樣中,本發明提供一種治療關節炎之醫藥 組成物,包括山竹(Garcinia mangostana L.)萃取物為活性成分。In one aspect of the invention, the invention provides a pharmaceutical composition for treating arthritis, comprising an extract of Mangosteen ( Garcinia mangostana L.) as an active ingredient.

在另一發明態樣中,本發明提供一種治療關節炎之醫藥組成物,包括下式(I)化合物或其鹽或酯為活性成分, 其中,R1 與R2 可相同或相異,分別為H或C1 -C6 烷基。In another aspect of the invention, the present invention provides a pharmaceutical composition for treating arthritis, comprising a compound of the following formula (I) or a salt or ester thereof as an active ingredient, Wherein R 1 and R 2 may be the same or different and are each H or a C 1 -C 6 alkyl group.

本發明更提供一種山竹萃取物之用途,用於製造治療關節炎之醫藥,以及一種式(I)化合物之用途,用於製造治療關節炎之醫藥。The present invention further provides a use of a mangosteen extract for the manufacture of a medicament for the treatment of arthritis, and a use of a compound of the formula (I) for the manufacture of a medicament for the treatment of arthritis.

在一發明態樣中,本發明提供一種調節細胞激素分泌之醫藥組成物,包括山竹(Garcinia mangostana L.)萃取物為活性成分。In one aspect of the invention, the invention provides a pharmaceutical composition for modulating cytokine secretion, comprising an extract of Mangosteen ( Garcinia mangostana L.) as an active ingredient.

在另一發明態樣中,本發明提供一種調節細胞激素分泌之醫藥組成物,包括式(I)化合物或其鹽或酯為活性成分,In another aspect of the invention, the present invention provides a pharmaceutical composition for regulating secretion of a cytokine, comprising a compound of the formula (I) or a salt or ester thereof as an active ingredient,

本發明更提供一種山竹萃取物之用途,用於製造調節細胞激素分泌之醫藥,以及一種式(I)化合物之用途,用於製造調節細胞激素分泌之醫藥。The present invention further provides a use of a mangosteen extract for the manufacture of a medicament for regulating secretion of a cytokine, and a use of a compound of the formula (I) for the manufacture of a medicament for regulating secretion of a cytokine.

本發明之具體實施詳細說明如下,然而以下的實施例僅用於進一步揭露本發明之技術內容,不應藉以限制本案的發明範疇。The specific embodiments of the present invention are described in detail below, but the following embodiments are only used to further disclose the technical content of the present invention, and should not limit the scope of the invention.

本發明之山竹萃取物係萃取自山竹植物體,較佳選用山竹果皮的部分。在本發明之一實施例中,選用山竹果皮乾燥物,經有機溶液萃取。前述有機溶液沒有特別限制,可以是C1 -C12 醇類,例如甲醇、乙醇、丙醇、異丙醇、丁醇、2-丁醇、戊醇、己醇、庚醇、辛醇、壬醇、癸醇、十一醇或十二醇等;或者芳香烴類,例如苯、甲苯或二甲苯。本發明之一實施例中使用乙醇水溶液萃取。乙醇水溶液的濃度可為10~90%,較佳可為20~80%,更佳可為50~75%。在本發明之一實施例中,使用加熱的乙醇水溶液萃取,加熱溫度為約80~85℃。在另一實施例中,在有機溶液溶液萃取之前,山竹乾燥物可先經由等重量的熱水處理,熱水的溫度約90℃以上,較佳介於95℃~100℃的範圍。在有機溶液萃取之前進行熱水處理,可事先除去山竹中具有顏色、黏性、吸濕等的雜質。上述萃取液經過乾燥之後,可獲得粗結晶,其中含有多種山竹素。上述之萃取方法,可進行一回,獲得粗萃取物或粗結晶,或重複數回,獲得較精純的山竹萃取物或結晶物。The mangosteen extract of the present invention is extracted from the mangosteen plant body, and the part of the mangosteen peel is preferably used. In an embodiment of the invention, the dried mangosteen peel is selected and extracted with an organic solution. The aforementioned organic solution is not particularly limited and may be a C 1 -C 12 alcohol such as methanol, ethanol, propanol, isopropanol, butanol, 2-butanol, pentanol, hexanol, heptanol, octanol, anthracene. Alcohol, decyl alcohol, undecyl alcohol or dodecyl alcohol, etc.; or aromatic hydrocarbons such as benzene, toluene or xylene. In one embodiment of the invention, an aqueous ethanol solution is used for extraction. The concentration of the aqueous ethanol solution may be from 10 to 90%, preferably from 20 to 80%, more preferably from 50 to 75%. In one embodiment of the invention, extraction is carried out using a heated aqueous solution of ethanol at a temperature of about 80 to 85 °C. In another embodiment, prior to the extraction of the organic solution solution, the dried mangosteen may be first treated with an equal weight of hot water having a temperature of about 90 ° C or higher, preferably between 95 ° C and 100 ° C. The hot water treatment before the extraction of the organic solution can remove impurities such as color, viscosity, moisture absorption, etc. in the mangosteen. After the above extract is dried, a crude crystal is obtained which contains various mangostin. The above extraction method can be carried out once to obtain a crude extract or crude crystal, or repeated several times to obtain a relatively pure mangosteen extract or crystal.

將上述萃取方法所得的山竹萃取物經矽膠管柱層析法(silicon chromatography)之後,以高速液相層析法(HPLC)分析,可獲得如下所示之式(I)化合物The mangosteen extract obtained by the above extraction method is subjected to high performance liquid chromatography (HPLC) analysis by means of silica chromatography to obtain a compound of the formula (I) shown below.

其中,R1 與R2 可相同或相異,分別為H或C1 -C6 烷基。Wherein R 1 and R 2 may be the same or different and are each H or a C 1 -C 6 alkyl group.

上述式(I)化合物中,當R1 為甲基、R2 為氫時,其為α-山竹素(mangostin);當R1 、R2 皆為甲基時,其為β-山竹素(mangostin);及當R1 、R2 皆為氫時,其為γ-山竹素(mangostin)。In the compound of the above formula (I), when R 1 is a methyl group and R 2 is hydrogen, it is α-mangostin; when both R 1 and R 2 are a methyl group, it is β-mancelin ( Mangostin); and when both R 1 and R 2 are hydrogen, it is γ-mangostin.

上述式(I)化合物可為鹽類或酯類,沒有特別限定,只要不影響式(I)化合物的治療關節炎及調節細胞激素分泌的功效者,皆可使用,例如鈉鹽、鉀鹽、鋰鹽、鎂鹽、鈣鹽、銨鹽、碳酸鹽、硝酸鹽、碳酸氫鹽、鹽酸鹽、硫酸鹽、磷酸鹽、或矽酸鹽等之鹽類;或者例如甲酯、乙酯、丙酯、丁酯、戊酯、乙酸甲酯、乙酸乙酯、甲酸丁酯、乙酸丁酯、戊酸丁酯、丙酸丁酯、丁酸甲酯、丁酸乙酯等之酯類。The compound of the above formula (I) may be a salt or an ester, and is not particularly limited as long as it does not affect the therapeutic effect of the compound of the formula (I) for treating arthritis and regulating cytokine secretion, such as sodium salt, potassium salt, a salt of a lithium salt, a magnesium salt, a calcium salt, an ammonium salt, a carbonate, a nitrate, a hydrogencarbonate, a hydrochloride, a sulfate, a phosphate, or a citrate; or, for example, a methyl ester, an ethyl ester, or a C Esters of esters, butyl esters, amyl esters, methyl acetate, ethyl acetate, butyl formate, butyl acetate, butyl valerate, butyl propionate, methyl butyrate, ethyl butyrate and the like.

本發明之醫藥組成物,除了式(I)化合物為活性成分外,可包括醫藥上可接受之賦形劑或其他添加劑,根據投藥方式及依該技術領域之慣例調配。投藥路徑可包括口服、經皮膚投藥、腹腔內投藥、靜脈內投藥、經鼻投藥、或眼部投藥等,較佳為口服。投藥劑量可根據患者年齡、體重、健康狀況、疾病種類、疾病的進展、患部等因素,由相關醫療人員依該技術領域中共通知識決定。本發明之醫藥組成物亦可單獨投藥或與其他藥劑共同投藥,投藥療程可依據藥學上例行方法實施。The pharmaceutical composition of the present invention may comprise, in addition to the active ingredient, a pharmaceutically acceptable excipient or other additive, depending on the mode of administration and according to the practice in the art. The route of administration may include oral administration, transdermal administration, intraperitoneal administration, intravenous administration, nasal administration, or ocular administration, and is preferably oral. The dosage can be determined by the relevant medical personnel according to the common knowledge in the technical field according to the patient's age, weight, health status, disease type, disease progression, and affected parts. The pharmaceutical composition of the present invention can also be administered alone or in combination with other agents, and the administration course can be carried out according to a pharmacy routine.

本發明所述之關節炎表示關節發炎的病症,然而發炎的症狀也可能發生在肌腱、韌帶、硬骨、軟骨、或肌肉組織。此述的關節炎可能來自於外在環境的引發,例如病毒或細菌感染造成的感染性關節炎(infectious arthritis),也可能因為免疫系統異常或基因變異所引起,例如風濕性關節炎、兒童期特異性關節炎(juvenile idiopathic arthritis)、紅斑性狼瘡(systemic lupus erythematosus)等。此述之關節炎尚包括退化性關節炎(osteoarthritis)、纖維肌痛症(fibromyalgia)、硬皮症(scleroderma)、脊椎關節炎(spondyloarthropathies)、痛風(gout)、風濕性多發性肌痛症(polymyalgia rheumatica)、多發性肌痛症(polymyositis)、乾癬性關節炎(psoriatic arthritis)、滑液囊炎(bursitis)、或肌腱炎(tendonitis)等。然而,本發明之關節炎不限於此述之疾病,任何病因造成的關節或相關組織的發炎皆包含於本發明所述之關節炎。The arthritis of the present invention means a condition in which the joint is inflamed, however, the symptoms of inflammation may also occur in tendons, ligaments, hard bones, cartilage, or muscle tissue. The arthritis described may be caused by an external environment, such as infectious arthritis caused by a viral or bacterial infection, or may be caused by abnormalities in the immune system or genetic variations, such as rheumatoid arthritis, childhood. Juvenile idiopathic arthritis, systemic lupus erythematosus, and the like. The arthritis described also includes osteoarthritis, fibromyalgia, scleroderma, spondyloarthropathies, gout, rheumatic polymyalgia ( Polymyalgia rheumatica), polymyositis, psoriatic arthritis, bursitis, or tendonitis. However, the arthritis of the present invention is not limited to the diseases described herein, and inflammation of joints or related tissues caused by any cause is included in the arthritis of the present invention.

造成關節炎的因素,多篇研究顯示與細胞激素有關,例如腫瘤壞死因子(tumor necrosis factor α,簡稱TNF-α)及介白素(interleukins,簡稱IL),例如IL-1、IL-4、IL-6等有關(Tracey D,et al.,Tumor necrosis factor antagonist mechanisms of action:a comprehensive review,Pharmacology & Therapeutics 117(2008)244-279.)。Tracey回顧多篇關節炎的研究指出核因子κ-B配體的受體活化物(receptor activator of nuclear factor κ-B ligand;RANKL)造成破骨的效應由TNF、IL-1、IL-6、IL-17及其他細胞激素所促進,並由干擾素IFNγ及介白素IL-4所抑制,在風濕性關節炎中RANKL對其受體的比例增加被認為是因為增加的破骨活性所造成(Tracey D,et al.,Tumor necrosis factor antagonist mechanisms of action:a comprehensive review,Pharmacology & Therapeutics 117(2008)244-279.)。因此,對於治療關節炎的候選藥物,可經由檢測該候選藥物是否造成生物體內細胞激素TNF-α、IL-4、IL-6的含量或分泌量降低,藉以評估其治療關節炎的效果。Factors that cause arthritis, many studies have shown that it is related to cytokines, such as tumor necrosis factor α (TNF-α) and interleukins (IL), such as IL-1, IL-4, IL-6 et al. (Tracey D, et al., Tumor necrosis factor antagonist mechanisms of action: a comprehensive review, Pharmacology & Therapeutics 117 (2008) 244-279.). Tracey recalls that several arthritis studies have indicated that the receptor effector of nuclear factor κ-B ligand (RANKL) causes osteoclast effect by TNF, IL-1, IL-6, It is promoted by IL-17 and other cytokines and is inhibited by interferon IFNγ and interleukin IL-4. The increase in the ratio of RANKL to its receptor in rheumatoid arthritis is thought to be caused by increased osteoclast activity. (Tracey D, et al., Tumor necrosis factor antagonist mechanisms of action: a comprehensive review, Pharmacology & Therapeutics 117 (2008) 244-279.). Therefore, for a drug candidate for treating arthritis, the effect of treating arthritis can be evaluated by detecting whether the drug candidate causes a decrease in the content or secretion amount of cytokines TNF-α, IL-4, IL-6 in the living body.

另有多篇研究建立關節炎的動物模式,例如Thorbecke等人使用膠原蛋白誘導的關節炎(collagen-induced arthritis;CIA)動物模式,顯示抑制TNF可明顯減緩關節炎的進展(Thorbecke G. J.,et al.,Involvement of endogenous tumor necrosis factor alpha and transforming growth factor beta during induction of collagen type II arthritis in mice.Proc Natl Acad Sci U S A 89,(1992)7375-7379.);或Mazzon等人使用鹿角膠誘導的掌腫脹(carrageennan-induced paw edema)模式,有效地評估在發炎過程中TNF-α扮演的角色(Mazzon E.,et al.,Effect of tumour necrosis factor-αreceptor 1 genetic deletion on carrageenan-induced acute inflammation:a comparison with etanercept,British Society for Immunology,Clinical and Experimental Immunology, 153(2008):136-149.)。此述文獻全文參照併入本申請案之內容。A number of other studies have established animal models of arthritis, such as Thorbecke et al. using collagen-induced arthritis (CIA) animal models, showing that inhibition of TNF significantly slows the progression of arthritis (Thorbecke GJ, et al Involvement of endogenous tumor necrosis factor alpha and transforming growth factor beta during induction of collagen type II arthritis in mice. Proc Natl Acad Sci USA 89, (1992) 7375-7379.); or Mazzon et al. The carrageennan-induced paw edema model effectively evaluates the role of TNF-α during inflammation (Mazzon E., et al., Effect of tumour necrosis factor-αreceptor 1 genetic deletion on carrageenan-induced acute inflammation:a Comparison with etanercept, British Society for Immunology, Clinical and Experimental Immunology, 153 (2008): 136-149.). The entire disclosure of this document is incorporated by reference in its entirety.

本發明人根據先前文獻建立之關節炎動物模式及關節炎細胞株,進行下列實施例,評估本發明之山竹萃取物及分離自該山竹萃取物的式(I)化合物或其鹽或酯作為治療關節炎之活性成分的潛力。在下列的細胞試驗及關節炎動物模式中,本發明之山竹萃取物及由該萃取物中分離的式(I)化合物或其鹽或酯明顯使細胞激素TNF-α、IL-4、IL-6的含量減少及減緩動物模式的關節炎症狀,呈現良好的治療關節炎效果,特別是α-山竹素、β-山竹素及γ-山竹素。而且由於山竹萃取物為農產廢棄物所提煉之產品,具有價廉、安全、可以口服的優點,在醫療應用上具有優勢。The present inventors conducted the following examples according to the arthritis animal model and the arthritis cell strain established in the prior literature, and evaluated the mangosteen extract of the present invention and the compound of the formula (I) or a salt or ester thereof isolated from the mangosteen extract as a treatment. The potential of active ingredients in arthritis. In the following cell tests and arthritic animal models, the mangosteen extract of the present invention and the compound of formula (I) or a salt or ester thereof isolated from the extract significantly occludes the cytokines TNF-α, IL-4, IL- The content of 6 is reduced and the arthritis symptoms of the animal model are slowed down, and the arthritis effect is well treated, especially α-mangostin, β-mangostin and γ-mangostin. Moreover, because mangosteen extract is a product refined from agricultural waste, it has the advantages of being cheap, safe, and can be taken orally, and has advantages in medical applications.

[實施例1]山竹萃取物之製造方法[Example 1] Method for producing mangosteen extract

(a)山竹萃取物:(a) Mangosteen extract:

山竹果皮乾燥物60公斤加入水600公升,加熱到90℃以上,並攪拌一小時洗淨,在加熱洗淨工程中除去顏色、黏性、吸濕等雜質。洗淨之果皮過濾後之殘渣,加入600公升之50%乙醇溶液,加熱至80~85℃攪拌一小時後過濾。濾液在常溫中放置過夜,析出粗結晶。過濾取得粗結晶。分離之粗結晶以10公升之50%乙醇洗淨之,在60℃下真空乾燥後得到粗山竹素結晶粉末2.4公斤。Mangosteen peel dry matter 60 kg is added to water 600 liters, heated to above 90 ° C, and stirred for one hour to wash, remove color, viscosity, moisture absorption and other impurities in the heating and washing process. The washed residue of the peel was added to a 600 liter 50% ethanol solution, heated to 80-85 ° C and stirred for one hour and then filtered. The filtrate was allowed to stand at room temperature overnight to precipitate a crude crystal. Filtration gave crude crystals. The separated crude crystals were washed with 10 liters of 50% ethanol, and vacuum dried at 60 ° C to obtain 2.4 kg of crude mangosteen crystal powder.

(b)山竹素之分離:(b) Separation of mangosteen:

將結晶粉末2.4公斤加入乙醇24公升,在常溫中攪拌一小時溶解,將此溶解液在常溫下一邊攪拌一邊慢慢滴入36公升之水中,繼續在80~85℃中攪拌一小時之後,繼續攪拌並慢慢冷卻至室溫。繼續在室溫中攪拌過夜使山竹素結晶析出,所得之結晶並以50%乙醇水溶液洗淨,並真空乾燥之,得到山竹素的組合物(含有α-山竹素75~85%,γ-山竹素7~15%)。2.4 kg of the crystal powder was added to 24 liters of ethanol, and stirred at room temperature for one hour to dissolve. The solution was slowly added dropwise to 36 liters of water while stirring at room temperature, and stirring was continued at 80 to 85 ° C for one hour, and then continued. Stir and slowly cool to room temperature. Continue to stir at room temperature overnight to crystallize the mangosteen, and crystallize it and wash it with 50% aqueous solution of ethanol, and dry it in vacuo to obtain a composition of mangostin (containing 75-85% of α-mangosin, γ-mangosteen 7 to 15%).

[實施例2]活體外(in vitro )細胞株之製備[Example 2] Preparation of in vitro cell line

U937細胞株U937 cell line

使用來自美國菌種保藏中心(ATCC)的人體骨髓白血球(myeloid leukemia)細胞株U937(Rockville,MD)。將該細胞株培養於37℃、5%CO2 、含10%胎牛血清(FCS)的RPMT 1640培養基中,維持在指數生長(exponential growth)狀態。為了進行分化誘導,將該細胞株以起始濃度為4x105 個細胞/ml,培養於含有50ng/ml PMA(Sigma)的T150培養瓶中24小時。之後移到未含PMA的相同培養基中,再培養48小時。之後以橡膠刮勺(rubber policeman)(Bellco Glass,Vineland,NJ)輕微刮除該培養瓶,收集細胞,用於以下實施例。A human myeloid leukemia cell line U937 (Rockville, MD) from the American Type Culture Collection (ATCC) was used. The cell strain was cultured in RPMT 1640 medium containing 5% CO 2 and 10% fetal calf serum (FCS) at 37 ° C, maintained in an exponential growth state. For differentiation induction, the cell strain was cultured in a T150 flask containing 50 ng/ml PMA (Sigma) at an initial concentration of 4 x 10 5 cells/ml for 24 hours. It was then transferred to the same medium without PMA and cultured for an additional 48 hours. The flask was then lightly scraped off with a rubber policeman (Bellco Glass, Vineland, NJ) and cells were collected for use in the following examples.

EL-4細胞株EL-4 cell line

使用來自美國菌種保藏中心(ATCC)的小鼠T淋巴瘤(murine T lymphoma)細胞株EL-4(Rockville,MD)。將該細胞株培養於37℃、5%CO2 、含10%胎牛血清(FCS)的RPMT 1640培養基中,使其維持在指數生長(exponential growth)狀態,收集細胞,用於以下實施例。A mouse T lymphoma cell line EL-4 (Rockville, MD) from the American Type Culture Collection (ATCC) was used. This cell strain was cultured in RPMT 1640 medium containing 5% CO 2 and 10% fetal bovine serum (FCS) at 37 ° C, maintained in an exponential growth state, and cells were collected for use in the following examples.

[實施例3]山竹萃取物對IL-4抑制作用及細胞毒性分析[Example 3] Inhibition of IL-4 by Mangosteen Extract and Analysis of Cytotoxicity

IL-4抑制作用IL-4 inhibition

在96孔盤中,植入1x104 個EL-4細胞,先與濃度分別為1.25μg/ml、2.5μg/ml、5μg/ml及10μg/ml的實施例1所得的山竹萃取物30μl於37℃下反應2小時,再在每孔加入20μl的PMA(1.5ng/well)及卡西黴素(Calcimycin)(A23187,Sigma)(15ng/well),在37℃、5%CO2 條件下培養細胞18小時。隔天,收集細胞培養液,利用ELISA(IL-4 duoset,R&D,Minneapolis,MN)檢測其IL-4含量。結果如表1及第1圖所示,以單獨含有PMA(phorbol 12-myristate 13-acetate,Sigma)及卡西黴素(Calcimycin)溶液的控制組為基礎,以百分比顯示IL-4分泌量的比率。In a 96-well plate, 1×10 4 EL-4 cells were implanted, and 30 μl of the mangosteen extract obtained in Example 1 at a concentration of 1.25 μg/ml, 2.5 μg/ml, 5 μg/ml, and 10 μg/ml, respectively, at 37 The reaction was carried out at ° C for 2 hours, and then 20 μl of PMA (1.5 ng/well) and calcimycin (A23187, Sigma) (15 ng/well) were added to each well, and cultured at 37 ° C under 5% CO 2 . The cells were 18 hours. On the next day, the cell culture medium was collected, and its IL-4 content was measured by ELISA (IL-4 duoset, R&D, Minneapolis, MN). The results are shown in Table 1 and Figure 1. Based on the control group containing PMA (phorbol 12-myristate 13-acetate, Sigma) and calcimycin solution alone, the IL-4 secretion was expressed as a percentage. ratio.

細胞毒性分析Cytotoxicity analysis

將上述實施例IL-4抑制作用試驗取出培養液後餘下之細胞,在37℃將1mg/ml的MTT(3-(4,5-dimethyl-thiazol-2-yl)2,5-diphenyltrazolium,Sigma)加入作用1小時,獲得的結晶物,加入100μl的DMSO,使該結晶物溶解。以ELISA Reader(Tecan Spectrafluor plus,Swizerland)測量該溶解結晶物的吸光度(OD560 )。結果如表1及第1圖所示,以單獨含有PMA及卡西黴素(Calcimycin)溶液的控制組為基礎,以百分比顯示細胞存活率。The IL-4 inhibitory test of the above example was taken out and the remaining cells were taken out, and 1 mg/ml of MTT (3-(4,5-dimethyl-thiazol-2-yl)2,5-diphenyltrazolium, Sigma was added at 37 °C. The crystals obtained were added for 1 hour, and 100 μl of DMSO was added to dissolve the crystals. The absorbance (OD 560 ) of the dissolved crystals was measured by ELISA Reader (Tecan Spectrafluor plus, Swizerland). The results are shown in Table 1 and Figure 1, and the cell viability was shown as a percentage based on a control group containing PMA alone and a calcimycin solution.

[實施例4]山竹萃取物對TNF-α抑制作用及細胞毒性分析[Example 4] Inhibition of TNF-α by Mangosteen Extract and Analysis of Cytotoxicity

TNF-α抑制作用TNF-α inhibition

在一96孔盤中,每一孔中平均分裝如實施例2分化的U937細胞1.6x105 個。另外根據下表2所示的配方,取實施例1所獲得之山竹萃取物與上述細胞混合,於37℃、5%CO2 下培養30分鐘,之後在各培養基中分別加入200ng/ml的脂多醣體溶液(lipopolysaccharide;LPS)(於磷酸緩衝生理食鹽水(PBS)中),再培養4小時。該培養基經分析套組(R&D system,Minneapolis,MN)收集,進行TNF-的分析。結果以GraFit分析計算,如表2及第2圖所示。表2的數值係來自以單獨含有LPS溶液(0.1μg/ml)的控制組為基礎,百分比顯示各濃度的TNF-α抑制作用。獲得IC50 為4.8μg/ml。In a 96-well plate, 1.6× 10 5 U937 cells differentiated as in Example 2 were equally dispensed in each well. Further, according to the formulation shown in Table 2 below, the mangosteen extract obtained in Example 1 was mixed with the above cells, and cultured at 37 ° C, 5% CO 2 for 30 minutes, and then 200 ng / ml of fat was added to each medium. A lipopolysaccharide (LPS) (in phosphate buffered saline (PBS)) was further cultured for 4 hours. The medium was collected by an analytical kit (R&D system, Minneapolis, MN) for analysis of TNF-. The results were calculated by GraFit analysis as shown in Table 2 and Figure 2. The values in Table 2 are based on a control group containing LPS solution alone (0.1 μg/ml), and the percentages show inhibition of TNF-α at various concentrations. The IC 50 was obtained at 4.8 μg/ml.

細胞毒性分析Cytotoxicity analysis

同實施例3之步驟,但根據表2配方進行,結果如表2及第2圖所示。同樣地,表2的數值係來自以單獨含有LPS溶液(0.1μg/ml)的控制組為基礎,百分比顯示各濃度的細胞存活率。The procedure of Example 3 was followed, but according to the formulation of Table 2, the results are shown in Table 2 and Figure 2. Similarly, the values in Table 2 are based on a control group containing LPS solution alone (0.1 μg/ml), and the percentages show cell viability at each concentration.

[實施例5]α,β,γ-山竹素(mangostin)的TNF-α抑制作用及細胞毒性[Example 5] TNF-α inhibition and cytotoxicity of α,β,γ-mangostin

選用實施例2所得經分化的U973細胞株,如實施例4之方法,分別測試濃度為3.1、6.3、12.5、25.0μg/ml的α-山竹素及β-山竹素、及濃度為7.5、15.0、30.0、60.0μg/ml的γ-山竹素之TNF-α抑制作用。結果如第3a-3c圖所示,α,β,γ-山竹素對TNF-α抑制作用的IC50 分別為5.5μg/ml、8.3μg/ml、4.8μg/ml。The differentiated U973 cell strain obtained in Example 2 was used, and the α-mancoside and β-mancoside at concentrations of 3.1, 6.3, 12.5, 25.0 μg/ml were tested as in Example 4, and the concentrations were 7.5, 15.0. TNF-α inhibition of γ-mangostin at 30.0 and 60.0 μg/ml. As a result, as shown in Fig. 3a-3c, the IC 50 of α,β,γ-mangostin inhibiting TNF-α was 5.5 μg/ml, 8.3 μg/ml, and 4.8 μg/ml, respectively.

[實施例6]山竹萃取物及α-山竹素對LPS誘導的小鼠血漿中TNF-α及IL-6抑制作用[Example 6] Inhibition of mannan extract and α-mangosin on TNF-α and IL-6 in mouse plasma induced by LPS

採用獲得自台灣國家實驗室動物中心(National Laboratory Animal Center,Taiwan)的BALA/c小鼠。將該小鼠飼養於室溫23±2℃、12小時開燈、12小時關燈、及40-70%空氣溼度下。該實驗動物可自由餵食固體食物及自來水。所有實驗動物守則及其飼養係根據工業技術研究院的實驗動物管理小組委員(IACUC,ITRI)及台灣農業局(Council of Agriculture,Taiwan)的規定。BALA/c mice obtained from the National Laboratory Animal Center (Taiwan) were used. The mice were housed at room temperature 23 ± 2 ° C, turned on for 12 hours, turned off for 12 hours, and 40-70% air humidity. The experimental animals were free to feed solid food and tap water. All experimental animal codes and their breeding are based on the provisions of the Laboratory Animal Management Subcommittee of the Industrial Technology Research Institute (IACUC, ITRI) and the Council of Agriculture (Taiwan).

上述雄BALA/c小鼠以6隻一群,分別腹腔投予實施例1的300mg/kg山竹萃取物及100mg/kg的α-山竹素30分鐘。之後,每一隻小鼠皆再腹腔注射1mg/kg LPS及Cremophore EL(BASF)(以下簡稱CrEL)(5%乙醇在30% CrEL中)。在LPS注射後1.5小時,收集小鼠血液於含有肝素的小管(heparinated eppendrof)中。分別測量該小鼠血漿中TNF-α及IL-6的分泌量(mouse TNF-αduoset及IL-4 duoset,R&D,Minneapolis,MN),結果如第4a-4b圖所示。The above male BALA/c mice were administered intraperitoneally with 300 mg/kg of mangosteen extract of Example 1 and 100 mg/kg of α-mangostin for 30 minutes, respectively. Thereafter, each mouse was intraperitoneally injected with 1 mg/kg LPS and Cremophore EL (BASF) (hereinafter referred to as CrEL) (5% ethanol in 30% CrEL). At 1.5 hours after LPS injection, mouse blood was collected from heparinated eppendrof. The secretion of TNF-α and IL-6 in the plasma of the mice (mouse TNF-αduoset and IL-4 duoset, R&D, Minneapolis, MN) were measured, and the results are shown in Fig. 4a-4b.

[實施例7]山竹萃取物在鹿角膠(carrageenan)誘導的大鼠後掌腫脹(paw edema)之動物模式[Example 7] Animal model of mangosteen extract in carrageenan-induced paw edema of rats

採用Long-Evan大鼠,來自台灣國家實驗室動物中心(National Laboratory Animal Center,Taiwan)。將上述動物飼養於室溫23±2℃、12小時開燈、12小時關燈、及40-70%空氣溼度下。該實驗動物可自由餵食固體食物及自來水。所有實驗動物守則及其飼養係根據工業技術研究院的實驗動物管理小組委員訓練(IACUC,ITRI)及台灣農業局(Council of Agriculture,Taiwan)的規定。Long-Evan rats were used from the National Laboratory Animal Center (Taiwan). The animals were housed at room temperature 23 ± 2 ° C, turned on for 12 hours, turned off for 12 hours, and 40-70% air humidity. The experimental animals were free to feed solid food and tap water. All experimental animal codes and their breeding are based on the provisions of the Experimental Animal Management Subcommittee Training of the Industrial Technology Research Institute (IACUC, ITRI) and the Council of Agriculture (Taiwan).

上述飼養的Long-Evan大鼠5隻一群,在實驗前18-20小時禁食,但可自由喝水。首先,測試組口服實施例1之山竹萃取物40mg/kg;控制組投予1% CMC(Carboxymethyl Cellulose)載劑;對照組投予50mg/kg。經1小時後,所有大鼠的左後腳掌注射鹿角(carrageenan)-生理食鹽水溶液。注射鹿角膠(carrageenan)的5個小時內,每小時犧牲1隻大鼠,以體積變化掃描器(plethysmometer)(PV-01,DR instrument,Taiwan)測量該鼠後腳掌的腫脹體積。Five of the above-mentioned Long-Evan rats were fasted for 18-20 hours before the experiment, but they were free to drink water. First, the test group was orally administered with Mangosteen extract 40 mg/kg; the control group was administered with 1% CMC (Carboxymethyl Cellulose) carrier; 50mg/kg. After 1 hour, the left hind paw of all rats was injected with carrageenan-physiological saline solution. Within 5 hours of injection of carrageenan, one rat was sacrificed per hour, and the swelling volume of the hind paw of the mouse was measured by a plethysmometer (PV-01, DR instrument, Taiwan).

對鹿角膠(carrageenan)發炎反應的抑制率根據下式計算:The inhibition rate of the inflammatory reaction of carrageenan is calculated according to the following formula:

抑制率(%)=(Nt-Nv)/Nv x 100Inhibition rate (%) = (Nt - Nv) / Nv x 100

Nt:測試群的後腳掌狀腫脹體積的淨變化;Nt: the net change in the palm-shaped swelling volume of the hind paw of the test group;

Nv:載劑控制群的後腳掌體積的淨變化。Nv: Net change in the volume of the hind paw of the vehicle control group.

結果如第5圖所示,在注射鹿角膠(carrageenan)的3、4、5個小時後,投予40mg/kg山竹萃取物群的抑制率分別達到24%、29%、29%。負百分比顯示測試群的抑制效果。結果數據以平均值±S.E.表示,P<0.05。As a result, as shown in Fig. 5, after 3, 4, and 5 hours of injection of carrageenan, the inhibition rates of the 40 mg/kg mangosteen extract group were 24%, 29%, and 29%, respectively. A negative percentage shows the inhibitory effect of the test group. Results data are expressed as mean ± S.E., P < 0.05.

[實施例8]山竹萃取物在膠原蛋白(Collagen)誘導的關節炎動物模式[Example 8] Mangosteen extract in collagen (Collagen)-induced arthritis animal model

採用Lewis大鼠,獲得自台灣國家實驗室動物中心(National Laboratory Animal Center,Taiwan)。將上述動物飼養於室溫23±2℃、12小時開燈、12小時關燈、及40-70%空氣溼度下。該實驗動物可自由餵食固體食物及自來水。所有實驗動物守則及其飼養係根據工業技術研究院的實驗動物管理小組委員訓練(IACUC,ITRI)及台灣農業局(Council of Agriculture,Taiwan)的規定。Lewis rats were obtained from the National Laboratory Animal Center (Taiwan). The animals were housed at room temperature 23 ± 2 ° C, turned on for 12 hours, turned off for 12 hours, and 40-70% air humidity. The experimental animals were free to feed solid food and tap water. All experimental animal codes and their breeding are based on the provisions of the Experimental Animal Management Subcommittee Training of the Industrial Technology Research Institute (IACUC, ITRI) and the Council of Agriculture (Taiwan).

對上述Lewis大鼠的尾巴根部皮下注射50μg的胎牛型II膠原蛋白(Collagen)(CII,Chronderx)乳化於完全弗氏輔劑(CFA,Sigma)中。同一天內,標記該大鼠的踝關節,作為腳掌體積測量的基礎。在上述的初級免疫後7天,全體大鼠再注射100μg的CII於不完全弗氏輔劑(IFA,Sigma)。將上述大鼠分為3群,於上述關節炎誘導日開始,每日分別口服投予100mg/kg的山竹萃取物、30mg/kg的、及1%CMC(Carboxymethyl Cellulose)溶液(載劑),直到實驗結束。在上述再注射CII之後,每日調查大鼠的關節炎發病,特徵為腳掌出現紅斑及/或腫脹。每週3次測量關節炎的發生率及嚴重性,以關節炎分級系統評估。整個實驗中每週兩次測量腳掌體積及體重。根據關節周圍組織的腫脹及紅斑程度,每一腳掌臨床上關節炎特徵可評分為0-4分數(score):0表示腳掌未腫脹及未出現紅斑;1表示腳掌出現輕微紅斑,或腳趾、手指、踝關節或腕關節的1個關節腫脹;2表示腳掌出現紅斑,及腳趾或手指上有2個以上關節腫脹,或踝關節或腕關節出現中度紅斑及腫脹;3表示腳掌加重腫脹,限制踝關節的使用,腳不能踏;4表示腳掌過度腫脹,踝關節僵硬。The tail of the Lewis rats was subcutaneously injected with 50 μg of fetal bovine type II collagen (Collagen) (CII, Chronderx) in a complete Freund's adjuvant (CFA, Sigma). On the same day, the ankle joint of the rat was marked as the basis for the measurement of the volume of the foot. Seven days after the primary immunization described above, all rats were reinjected with 100 μg of CII in incomplete Freund's adjuvant (IFA, Sigma). The above-mentioned rats were divided into three groups, and 100 mg/kg of mangosteen extract and 30 mg/kg were orally administered daily on the day of induction of the above-mentioned arthritis. And 1% CMC (Carboxymethyl Cellulose) solution (carrier) until the end of the experiment. After the above-mentioned reinjection of CII, the onset of arthritis in the rats was investigated daily, characterized by erythema and/or swelling of the soles of the feet. The incidence and severity of arthritis were measured 3 times a week and assessed by the arthritis grading system. Foot volume and body weight were measured twice a week throughout the experiment. According to the swelling of the tissues around the joints and the degree of erythema, the clinical arthritis characteristics of each foot can be scored as 0-4 scores: 0 means that the soles of the feet are not swollen and no erythema is present; 1 means that the feet have slight erythema, or toes, fingers One joint of the ankle or wrist joint is swollen; 2 indicates erythema on the sole of the foot, and more than 2 joints on the toe or finger are swollen, or moderate erythema and swelling in the ankle or wrist joint; 3 indicates swelling of the sole of the foot, limiting The use of the ankle joint, the foot can not step; 4 indicates excessive swelling of the sole of the foot, and the ankle joint is stiff.

16為每一動物的組合性關節炎分數中的最大分數(參考Rosloniec,EF,Cremer,M,Kang,A and Myers,LK. Collagen-Induced Arthritis. Current protocols in Immunology(1996)15.5.1-15.5.24.)。16 is the maximum score in the combined arthritis score for each animal (Ref. Rosloniec, EF, Cremer, M, Kang, A and Myers, LK. Collagen-Induced Arthritis. Current protocols in Immunology (1996) 15.5.1-15.5 .twenty four.).

上述的腳掌體積以體積變化掃描器(plethysmometer),以每一隻大鼠的平均體積表示。測量體重在0.1g的精密平衡儀(METTLLER TOLDEO,PB1501)。The above-mentioned foot volume is expressed as a volume change plethysmometer in terms of the average volume of each rat. A precision balancer (METTLLER TOLDEO, PB1501) weighing 0.1 g was measured.

結果如第6圖所示。數據以平均值±SEM表示,以Student’s t-test分析不同群間的差異,與載劑處理群相比。P<0.05認為具有統計上顯著性。The result is shown in Figure 6. Data are presented as mean ± SEM, and differences between groups were analyzed by Student's t-test compared to vehicle treated groups. P < 0.05 was considered to be statistically significant.

由第6圖的結果顯示,首先出現肉眼可見的關節炎特徵在免疫後第9天,載劑處理群則在第19天達到最大關節炎分數及腳掌腫脹,之後逐漸消退。山竹萃取物的處理群的發病日晚載劑處理組1天。載劑處理群的全體發病率在第12天,山竹萃取物的處理群在第14天僅達到67%直到實驗結束。Celebrex處理群由第14天的發病率為86%,直到實驗結束。The results from Fig. 6 show that the first visible arthritis characteristic on the 9th day after immunization, the carrier treatment group reached the maximum arthritis score and swelling of the paw on the 19th day, and then gradually subsided. The treatment group of the mangosteen extract was treated with the carrier treatment group for 1 day. The overall incidence of the vehicle treatment group was on the 12th day, and the treatment group of the mangosteen extract reached only 67% on the 14th day until the end of the experiment. Celebrex The incidence of the treatment group from day 14 was 86% until the end of the experiment.

由實驗結果得知,山竹萃取物的處理群降低膠原蛋白誘導的關節炎模式的發病率。載劑處理群的關節炎嚴重性平均分數為6.88,山竹萃取物的處理群則降低為3.50,Celebrex處理群為3.0。腳掌體積的變化顯示與分數評分的趨勢相近。It is known from the experimental results that the treatment group of mangosteen extract reduces the incidence of collagen-induced arthritis patterns. The average score of arthritis severity of the carrier treatment group was 6.88, and the treatment group of mangosteen extract was reduced to 3.50, Celebrex. The processing group is 3.0. The change in the volume of the foot shows a similar trend to the score.

雖然本發明已以較佳實施例揭露如上,然其並非用以限定本發明,任何熟悉此項技藝者,在不脫離本發明之精神和範圍內,當可做些許更動與潤飾,因此本發明之保護範圍當視後附之申請專利範圍所界定者為準。While the present invention has been described in its preferred embodiments, the present invention is not intended to limit the invention, and the present invention may be modified and modified without departing from the spirit and scope of the invention. The scope of protection is subject to the definition of the scope of the patent application.

第1圖顯示山竹萃取物的活體外(in vitro )抑制IL-4分泌的效應及細胞毒性。Figure 1 shows the effect of the mangosteen extract on inhibiting IL-4 secretion and cytotoxicity in vitro .

第2圖顯示山竹萃取物的活體外(in vitro) 抑制TNF-α分泌的效應及細胞毒性。Figure 2 shows the effect of cynomolgus extract on the inhibition of TNF-α secretion and cytotoxicity in vitro .

第3a圖顯示α-山竹素(α-mangostin)活體外(in vitro )抑制TNF-α分泌的效應及細胞毒性;Figure 3a shows the effect of α-mangostin inhibiting TNF-α secretion and cytotoxicity in vitro ;

第3b圖顯示顯示β-山竹素(β-mangostin)活體外(in vitro )抑制TNF-α分泌的效應及細胞毒性山竹素;及Figure 3b shows the effect of β-mangostin inhibiting TNF-α secretion and cytotoxic mangostin in vitro ;

第3c圖顯示 -山竹素( -mangostin)活體外(in vitro )抑制TNF-α分泌的效應及細胞毒性。Figure 3c shows that the effect of mangostin (-mangostin) inhibiting TNF-α secretion and cytotoxicity in vitro .

第4a圖顯示山竹萃取物及α-山竹素在脂多醣體(LPS)誘導的小鼠血漿中抑制TNF-α的效應;及Figure 4a shows the effect of mangosteen extract and α-mangosin inhibiting TNF-α in lipopolysaccharide (LPS)-induced mouse plasma;

第4b圖顯示山竹萃取物及α-山竹素在脂多醣體(LPS)誘導的小鼠血漿中抑制IL-6分泌的效應。Figure 4b shows the effect of mangosteen extract and α-mangosin inhibiting IL-6 secretion in lipopolysaccharide (LPS)-induced mouse plasma.

第5圖顯示在鹿角膠(carrageenen)誘導的大鼠掌腫脹(paw edema)模式中,山竹萃取物對掌腫脹的抑制作用。Figure 5 shows the inhibitory effect of mangosteen extract on palm swelling in a carrageenen-induced paw edema model.

第6圖顯示在膠原蛋白(collegan)誘導的關節炎動物模式中,山竹萃取物對關節炎患部的抑制作用。Fig. 6 shows the inhibitory effect of mangosteen extract on the affected part of arthritis in a collagen-induced arthritis animal model.

Claims (2)

一種下式(I)化合物之用途,用於製造調節細胞激素分泌之醫藥, [式中,R1 與R2 皆為甲基]其中,該細胞激素包括介白素(interleukins)或腫瘤壞死因子(TNF-α)。Use of a compound of the following formula (I) for the manufacture of a medicament for regulating the secretion of cytokines, [wherein, both R 1 and R 2 are a methyl group] wherein the cytokine includes interleukins or tumor necrosis factor (TNF-α). 如申請專利範圍第1項所述之用途,其中式(I)化合物分離自山竹(Garcinia mangostana L.)。The use according to claim 1, wherein the compound of formula (I) is isolated from Mangosteen ( Garcinia mangostana L.).
TW98145264A 2009-12-28 2009-12-28 Use of modulating secretion of cytokines TWI435727B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
TW98145264A TWI435727B (en) 2009-12-28 2009-12-28 Use of modulating secretion of cytokines

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
TW98145264A TWI435727B (en) 2009-12-28 2009-12-28 Use of modulating secretion of cytokines

Publications (2)

Publication Number Publication Date
TW201121556A TW201121556A (en) 2011-07-01
TWI435727B true TWI435727B (en) 2014-05-01

Family

ID=45045659

Family Applications (1)

Application Number Title Priority Date Filing Date
TW98145264A TWI435727B (en) 2009-12-28 2009-12-28 Use of modulating secretion of cytokines

Country Status (1)

Country Link
TW (1) TWI435727B (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI472618B (en) * 2014-01-15 2015-02-11 Ind Tech Res Inst Methods for enhancing stem cells differentiating into insulin-producing cells
CN112426516A (en) * 2020-12-02 2021-03-02 上海楷达生物科技有限公司 Composition for treating arthritis and preparation method thereof
CA3223022A1 (en) * 2021-11-05 2023-05-11 Huan-yuan CHEN Use of mangosteen fruit shell extract in the preparation of a medicament for treating psoriasis

Also Published As

Publication number Publication date
TW201121556A (en) 2011-07-01

Similar Documents

Publication Publication Date Title
US7807711B2 (en) Medicinal acidic cannabinoids
JP6290531B2 (en) Plectranthus amboinix fraction with anti-arthritic activity
Liu et al. Effects of taurochenodeoxycholic acid on adjuvant arthritis in rats
EA014070B1 (en) Use of a herbal composition for the treatment of inflammatory disorders
WO2018082568A1 (en) Extracts of andrographis paniculata, methods for preparation and use thereof
JPH085780B2 (en) Osteoarthritis treatment
WO2021249420A1 (en) Use of kadsura heteroclita (roxb.) craib agent in preparation of medicament for resisting rheumatoid arthritis
TWI320714B (en) Plant extracts for the treatment of rheumatoid arthritis
CN1566124A (en) Cornel extract and use thereof
CA3219768A1 (en) Pharmaceutical composition for treating rheumatoid arthritis and preparation method therefor
TWI435727B (en) Use of modulating secretion of cytokines
WO2013078764A1 (en) Applications of 1β-hydroxy isoalantolactone in preparing medicines against rheumatoid arthritis
WO2008047880A1 (en) Therapeutic agent for rheumatoid arthritis
CA3116793C (en) Medical use of anemoside b4 against acute gouty arthritis
CN116440110B (en) Medical application of tolypic acid and derivative thereof in preparation of medicines for treating immune and inflammation related diseases
CN109419787B (en) Application of abietane diterpenoid compound
WO2022178967A1 (en) Pharmaceutical composition of forskolin-isoforskolin and pentacyclic triterpenoid compound, and application thereof
CN116019813A (en) Application of Vesatolimod in preparation of medicines for preventing and/or treating central nervous system diseases
CN114699410A (en) Application of cepharanthine in preparing medicine for treating rheumatoid arthritis
CN108451949B (en) Application of paeoniflorin metabolite I in preparation of colitis treatment drug
CN116850180B (en) Application of cork xanthoxylin in preparing medicament for treating rheumatoid arthritis
TWI535443B (en) Combination of lupeol acetate and curcumin used for the treatment or prevention of activated osteoclast precursor related diseases
RU2784896C2 (en) Medical use of anemoside b4 against acute gouty arthritis
CN109966299B (en) Application of indole alkaloid in treating allergic rhinitis and pharmaceutical composition thereof
KR20140137557A (en) Anti-inflammation composition including artemisia extract and anthriscus sylvestris extract